Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Int J Mol Sci ; 25(11)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38892372

ABSTRACT

Organochlorine pesticides (OCPs) are a class of environmentally persistent and bioaccumulative pollutants. Among these, ß-hexachlorocyclohexane (ß-HCH) is a byproduct of lindane synthesis, one of the most worldwide widespread pesticides. ß-HCH cellular mechanisms inducing chemical carcinogenesis correspond to many of those inducing chemoresistance, in particular, by the activation of signal transducer and activator of transcription 3 (STAT3) signaling pathways. For this purpose, four cell lines, representative of breast, lung, prostate, and hepatocellular cancers, were treated with ß-HCH, specific tyrosine kinase inhibitors (TKIs), and a STAT3 inhibitor. All cell samples were analyzed by a viability assay, immunoblotting analysis, a wound-healing assay, and a colony formation assay. The results show that ß-HCH reduces the efficacy of TKIs. The STAT3 protein, in this context, plays a central role. In fact, by inhibiting its activity, the efficacy of the anticancer drug is restored. Furthermore, this manuscript aimed to draw the attention of the scientific and socio-healthcare community to the issue of prolonged exposure to contaminants and their impact on drug efficacy.


Subject(s)
Antineoplastic Agents , Hexachlorocyclohexane , Protein Kinase Inhibitors , STAT3 Transcription Factor , Signal Transduction , STAT3 Transcription Factor/metabolism , Humans , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Cell Line, Tumor , Hexachlorocyclohexane/pharmacology , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Neoplasms/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects
2.
Int J Mol Sci ; 24(17)2023 Aug 26.
Article in English | MEDLINE | ID: mdl-37686085

ABSTRACT

The protein disulfide isomerase A3 (PDIA3) is directly or indirectly involved in various physiopathological processes and participates in cancer initiation, progression and chemosensitivity. However, little is known about its involvement in glioblastoma. To obtain specific information, we performed cellular experiments in the T98G and U-87 MG glioblastoma cell lines to evaluate the role of PDIA3. The loss of PDIA3 functions, either through inhibition or silencing, reduced glioblastoma cells spreading by triggering cytotoxic phenomena. PDIA3 inhibition led to a redistribution of PDIA3, resulting in the formation of protein aggregates visualized through immunofluorescence staining. Concurrently, cell cycle progression underwent arrest at the G1/S checkpoint. After PDIA3 inhibition, ROS-independent DNA damage and the activation of the repair system occurred, as evidenced by the phosphorylation of H2A.X and the overexpression of the Ku70 protein. We also demonstrated through a clonogenic assay that PDIA3 inhibition could increase the chemosensitivity of T98G and U-87 MG cells to the approved glioblastoma drug temozolomide (TMZ). Overall, PDIA3 inhibition induced cytotoxic effects in the analyzed glioblastoma cell lines. Although further in vivo studies are needed, the results suggested PDIA3 as a novel therapeutic target that could also be included in already approved therapies.


Subject(s)
Glioblastoma , Humans , Glioblastoma/drug therapy , Protein Disulfide-Isomerases/genetics , Temozolomide/pharmacology , Phosphorylation , Biological Assay
3.
Int J Mol Sci ; 24(3)2023 Feb 03.
Article in English | MEDLINE | ID: mdl-36769334

ABSTRACT

In the present study, we used a mouse model of Alzheimer's disease (AD) (3×Tg-AD mice) to longitudinally analyse the expression level of PDIA3, a protein disulfide isomerase and endoplasmic reticulum (ER) chaperone, in selected brain limbic areas strongly affected by AD-pathology (amygdala, entorhinal cortex, dorsal and ventral hippocampus). Our results suggest that, while in Non-Tg mice PDIA3 levels gradually reduce with aging in all brain regions analyzed, 3×Tg-AD mice showed an age-dependent increase in PDIA3 levels in the amygdala, entorhinal cortex, and ventral hippocampus. A significant reduction of PDIA3 was observed in 3×Tg-AD mice already at 6 months of age, as compared to age-matched Non-Tg mice. A comparative immunohistochemistry analysis performed on 3×Tg-AD mice at 6 (mild AD-like pathology) and 18 (severe AD-like pathology) months of age showed a direct correlation between the cellular level of Aß and PDIA3 proteins in all the brain regions analysed, even if with different magnitudes. Additionally, an immunohistochemistry analysis showed the presence of PDIA3 in all post-mitotic neurons and astrocytes. Overall, altered PDIA3 levels appear to be age- and/or pathology-dependent, corroborating the ER chaperone's involvement in AD pathology, and supporting the PDIA3 protein as a potential novel therapeutic target for the treatment of AD.


Subject(s)
Alzheimer Disease , Mice , Animals , Alzheimer Disease/metabolism , Mice, Transgenic , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Brain/metabolism , Mice, Inbred Strains , Disease Models, Animal , Amyloid beta-Peptides/metabolism , tau Proteins/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism
4.
J Neurochem ; 162(4): 322-336, 2022 08.
Article in English | MEDLINE | ID: mdl-35699375

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder whose main pathological hallmark is the accumulation of Amyloid-ß peptide (Aß) in the form of senile plaques. Aß can cause neurodegeneration and disrupt cognitive functions by several mechanisms, including oxidative stress. ERp57 is a protein disulfide isomerase involved in the cellular stress response and known to be present in the cerebrospinal fluid of normal individuals as a complex with Aß peptides, suggesting that it may be a carrier protein which prevents aggregation of Aß. Although several studies show ERp57 involvement in neurodegenerative diseases, no clear mechanism of action has been identified thus far. In this work, we gain insights into the interaction of Aß with ERp57, with a special focus on the contribution of ERp57 to the defense system of the cell. Here, we show that recombinant ERp57 directly interacts with the Aß25-35 fragment in vitro with high affinity via two in silico-predicted main sites of interaction. Furthermore, we used human neuroblastoma cells to show that short-term Aß25-35 treatment induces ERp57 decrease in intracellular protein levels, different intracellular localization, and ERp57 secretion in the cultured medium. Finally, we demonstrate that recombinant ERp57 counteracts the toxic effects of Aß25-35 and restores cellular viability, by preventing Aß25-35 aggregation. Overall, the present study shows that extracellular ERp57 can exert a protective effect from Aß toxicity and highlights it as a possible therapeutic tool in the treatment of AD.


Subject(s)
Alzheimer Disease , Neurons , Peptide Fragments , Protein Disulfide-Isomerases , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Humans , Neurons/metabolism , Peptide Fragments/metabolism , Protein Disulfide-Isomerases/metabolism
5.
Cell Mol Biol Lett ; 27(1): 12, 2022 Feb 02.
Article in English | MEDLINE | ID: mdl-35109791

ABSTRACT

The ERp57/PDIA3 protein is a pleiotropic member of the PDIs family and, although predominantly located in the endoplasmic reticulum (ER), has indeed been found in other cellular compartments, such as the nucleus or the cell membrane. ERp57/PDIA3 is an important research target considering it can be found in various subcellular locations. This protein is involved in many different physiological and pathological processes, and our review describes new data on its functions and summarizes some ligands identified as PDIA3-specific inhibitors.


Subject(s)
Endoplasmic Reticulum , Protein Disulfide-Isomerases , Cell Membrane/metabolism , Cell Nucleus/metabolism , Endoplasmic Reticulum/metabolism , Protein Disulfide-Isomerases/metabolism
6.
Molecules ; 27(12)2022 Jun 14.
Article in English | MEDLINE | ID: mdl-35744943

ABSTRACT

Cancer is a disease of high mortality, and its prevalence has increased steadily in the last few years. However, during the last decade, the development of modern chemotherapy schemes, new radiotherapy techniques, targeted therapies and immunotherapy has brought new hope in the treatment of these diseases. Unfortunately, cancer therapies are also associated with frequent and, sometimes, severe adverse events. Ascorbate (ascorbic acid or vitamin C) is a potent water-soluble antioxidant that is produced in most mammals but is not synthesised endogenously in humans, which lack enzymes for its synthesis. Ascorbate has antioxidant effects that correspond closely to the dose administered. Interestingly, this natural antioxidant induces oxidative stress when given intravenously at a high dose, a paradoxical effect due to its interactions with iron. Importantly, this deleterious property of ascorbate can result in increased cell death. Although, historically, ascorbate has been reported to exhibit anti-tumour properties, this effect has been questioned due to the lack of available mechanistic detail. Recently, new evidence has emerged implicating ferroptosis in several types of oxidative stress-mediated cell death, such as those associated with ischemia-reperfusion. This effect could be positively modulated by the interaction of iron and high ascorbate dosing, particularly in cell systems having a high mitotic index. In addition, it has been reported that ascorbate may behave as an adjuvant of favourable anti-tumour effects in cancer therapies such as radiotherapy, radio-chemotherapy, chemotherapy, immunotherapy, or even in monotherapy, as it facilitates tumour cell death through the generation of reactive oxygen species and ferroptosis. In this review, we provide evidence supporting the view that ascorbate should be revisited to develop novel, safe strategies in the treatment of cancer to achieve their application in human medicine.


Subject(s)
Antioxidants , Neoplasms , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Antioxidants/therapeutic use , Ascorbic Acid/pharmacology , Ascorbic Acid/therapeutic use , Humans , Iron/metabolism , Mammals/metabolism , Neoplasms/drug therapy , Oxidative Stress , Reactive Oxygen Species/metabolism
7.
Int J Mol Sci ; 22(11)2021 May 29.
Article in English | MEDLINE | ID: mdl-34072471

ABSTRACT

Organochlorine pesticides constitute the majority of the total environmental pollutants, and a wide range of compounds have been found to be carcinogenic to humans. Among all, growing interest has been focused on ß-hexachlorocyclohexane (ß-HCH), virtually the most hazardous and, at the same time, the most poorly investigated member of the hexachlorocyclohexane family. Considering the multifaceted biochemical activities of ß-HCH, already established in our previous studies, the aim of this work is to assess whether ß-HCH could also trigger cellular malignant transformation toward cancer development. For this purpose, experiments were performed on the human normal bronchial epithelium cell line BEAS-2B exposed to 10 µM ß-HCH. The obtained results strongly support the carcinogenic potential of ß-HCH, which is achieved through both non-genotoxic (activation of oncogenic signaling pathways and proliferative activity) and indirect genotoxic (ROS production and DNA damage) mechanisms that significantly affect cellular macroscopic characteristics and functions such as cell morphology, cell cycle profile, and apoptosis. Taking all these elements into account, the presented study provides important elements to further characterize ß-HCH, which appears to be a full-fledged carcinogenic agent.


Subject(s)
Carcinogens/pharmacology , Cell Transformation, Neoplastic/chemically induced , Epithelial Cells/drug effects , Epithelial Cells/pathology , Hexachlorocyclohexane/pharmacology , Respiratory Mucosa/drug effects , Respiratory Mucosa/pathology , Apoptosis/drug effects , Biomarkers , Biomarkers, Tumor , Cell Cycle/drug effects , Cell Line , Cell Proliferation , Cells, Cultured , Epithelial Cells/metabolism , Gene Expression , Hexachlorocyclohexane/adverse effects , Humans , Reactive Oxygen Species/metabolism , Respiratory Mucosa/metabolism , Signal Transduction/drug effects
8.
Molecules ; 26(23)2021 Nov 25.
Article in English | MEDLINE | ID: mdl-34885717

ABSTRACT

The ß-isomer of hexachlorocyclohexane (ß-HCH) is a globally widespread pollutant that embodies all the physicochemical characteristics of organochlorine pesticides, constituting an environmental risk factor for a wide range of noncommunicable diseases. Previous in vitro studies from our group disclosed the carcinogenic potential of ß-HCH, which contributes to neoplastic transformation by means of multifaceted intracellular mechanisms. Considering the positive evidence regarding the protective role of natural bioactive compounds against pollution-induced toxicity, micronutrients from olive and tomato endowed with the capability of modulating ß-HCH cellular targets were tested. For this purpose, the solution obtained from a patented food supplement (No. EP2851080A1), referred to as Tomato and Olive Bioactive Compounds (TOBC), was administered to the androgen-sensitive prostate cancer cells LNCaP and different biochemical and cellular assays were performed to evaluate its efficiency. TOBC shows a dose-dependent significant chemoprotection by contrasting ß-HCH-induced intracellular responses such as STAT3 and AhR activation, disruption of AR signaling, antiapoptotic and proliferative activity, and increase in ROS production and DNA damage. These experimental outcomes identified TOBC as a suitable functional food to be included in a diet regimen aimed at defending cells from ß-HCH negative effects, recommending the development of tailored enriched formulations for exposed individuals.


Subject(s)
Phytochemicals/pharmacology , Prostatic Neoplasms/diet therapy , Receptors, Androgen/genetics , STAT3 Transcription Factor/genetics , Androgens/metabolism , Cell Proliferation/drug effects , DNA Damage/drug effects , Environmental Pollutants/toxicity , Gene Expression Regulation, Neoplastic/drug effects , Hexachlorocyclohexane/toxicity , Humans , Solanum lycopersicum/chemistry , Male , Micronutrients/chemistry , Micronutrients/pharmacology , Olea/chemistry , Phytochemicals/chemistry , Prostatic Neoplasms/chemically induced , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Reactive Oxygen Species/chemistry , Risk Factors , Signal Transduction/drug effects
9.
Int J Mol Sci ; 21(21)2020 Nov 03.
Article in English | MEDLINE | ID: mdl-33153019

ABSTRACT

The glioblastoma (GB) microenvironment includes cells of the innate immune system identified as glioma-associated microglia/macrophages (GAMs) that are still poorly characterized. A potential role on the mechanisms regulating GAM activity might be played by the endoplasmic reticulum protein ERp57/PDIA3 (protein disulfide-isomerase A3), the modulation of which has been reported in a variety of cancers. Moreover, by using The Cancer Genome Atlas database, we found that overexpression of PDIA3 correlated with about 55% reduction of overall survival of glioma patients. Therefore, we analyzed the expression of ERp57/PDIA3 using specimens obtained after surgery from 18 GB patients. Immunohistochemical analysis of tumor samples revealed ERp57/PDIA3 expression in GB cells as well as in GAMs. The ERp57/PDIA3 levels were higher in GAMs than in the microglia present in the surrounding parenchyma. Therefore, we studied the role of PDIA3 modulation in microglia-glioma interaction, based on the ability of conditioned media collected from human GB cells to induce the activation of microglial cells. The results indicated that reduced PDIA3 expression/activity in GB cells significantly limited the microglia pro-tumor polarization towards the M2 phenotype and the production of pro-inflammatory factors. Our data support a role of PDIA3 expression in GB-mediated protumor activation of microglia.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Macrophage Activation/genetics , Microglia/physiology , Protein Disulfide-Isomerases/genetics , Tumor Microenvironment/genetics , Adult , Aged , Aged, 80 and over , Brain Neoplasms/genetics , Cells, Cultured , Cohort Studies , Disease Progression , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Humans , Macrophages/metabolism , Macrophages/pathology , Macrophages/physiology , Male , Microglia/metabolism , Microglia/pathology , Middle Aged , Protein Disulfide-Isomerases/metabolism
10.
Int J Mol Sci ; 21(2)2020 Jan 17.
Article in English | MEDLINE | ID: mdl-31963614

ABSTRACT

Doxorubicin represents a valuable choice for different cancers, although the severe side effects occurring at the high effective dose limits its clinical use. In the present study, potential strategies to potentiate low-dose doxorubicin efficacy, including a metronomic schedule, characterized by a short and repeated exposure to the anticancer drug, and the combination with the natural chemosensitizing sesquiterpenes ß-caryophyllene and ß-caryophyllene oxide, were assessed in human hepatoma HepG2 cells. The involvement of P-glycoprotein (P-gp) in the HepG2-chemosensitization to doxorubicin was evaluated. Also, the direct interaction of caryophyllene sesquiterpenes with P-gp was characterized by molecular docking and dynamic simulation studies. A metronomic schedule allowed us to enhance the low-dose doxorubicin cytotoxicity and the combination with caryophyllane sesquiterpenes further potentiated this effect. Also, an increased intracellular accumulation of doxorubicin and rhodamine 123 induced by caryophyllane sesquiterpenes was found, thus suggesting their interference with P-gp function. A lowered expression of P-gp induced by the combinations, with respect to doxorubicin alone, was observed too. Docking studies found that the binding site of caryophyllane sesquiterpene was next to the ATP binding domain of P-gp and that ß-caryophyllene possessed the stronger binding affinity and higher inhibition potential calculated by MM-PBSA. Present findings strengthen our hypothesis about the potential chemosensitizing power of caryophyllane sesquiterpenes and suggest that combining a chemosensitizer and a metronomic schedule can represent a suitable strategy to overcome drawbacks of doxorubicin chemotherapy while exploiting its powerful activity.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Apoptosis , Carcinoma, Hepatocellular/pathology , Doxorubicin/pharmacology , Liver Neoplasms/pathology , Polycyclic Sesquiterpenes/chemistry , Sesquiterpenes/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Antibiotics, Antineoplastic/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Computer Simulation , Dose-Response Relationship, Drug , Humans , In Vitro Techniques , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Tumor Cells, Cultured
11.
Int J Mol Sci ; 20(8)2019 Apr 12.
Article in English | MEDLINE | ID: mdl-31013746

ABSTRACT

STAT3 is an oncoprotein overexpressed in different types of tumors, including prostate cancer (PCa), and its activity is modulated by a variety of post-translational modifications (PTMs). Prostate cancer represents the most common cancer diagnosed in men, and each phase of tumor progression displays specific cellular conditions: inflammation is predominant in tumor's early stage, whereas oxidative stress is typical of clinically advanced PCa. The aim of this research is to assess the correspondence between the stimulus-specificity of STAT3 PTMs and definite STAT3-mediated transcriptional programs, in order to identify new suitable pharmacological targets for PCa treatment. Experiments were performed on less-aggressive LNCaP and more aggressive DU-145 cell lines, simulating inflammatory and oxidative-stress conditions. Cellular studies confirmed pY705-STAT3 as common denominator of all STAT3-mediated signaling. In addition, acK685-STAT3 was found in response to IL-6, whereas glutC328/542-STAT3 and pS727-STAT3 occurred upon tert-butyl hydroperoxyde (tBHP) treatment. Obtained results also provided evidence of an interplay between STAT3 PTMs and specific protein interactors such as P300 and APE1/Ref-1. In accordance with these outcomes, mRNA levels of STAT3-target genes seemed to follow the differing STAT3 PTMs. These results highlighted the role of STAT3 and its PTMs as drivers in the progression of PCa.


Subject(s)
Prostatic Neoplasms/metabolism , Protein Processing, Post-Translational , STAT3 Transcription Factor/metabolism , Signal Transduction , Biomarkers , Gene Expression Regulation, Neoplastic , Humans , Male , Mitochondria/genetics , Mitochondria/metabolism , Prostatic Neoplasms/genetics , Transcription, Genetic
12.
Eur J Nutr ; 57(4): 1485-1498, 2018 Jun.
Article in English | MEDLINE | ID: mdl-28314964

ABSTRACT

PURPOSE: Chronic exposure to stress may represent a risk factor for developing metabolic and eating disorders, mostly driven by the overconsumption of easily accessible energy-dense palatable food, although the mechanisms involved remain still unclear. In this study, we used an ethologically oriented murine model of chronic stress caused by chronic psychosocial defeat (CPD) to investigate the effects of unrestricted access to a palatable high fat diet (HFD) on food intake, body weight, energy homeostasis, and expression of different brain neuropeptides. Our aim was to shed light on the mechanisms responsible for body weight and body composition changes due to chronic social stress. METHODS: In our model of subordinate (defeated), mice (CPD) cohabitated in constant sensory contact with dominants, being forced to interact on daily basis, and were offered ad libitum access either to an HFD or to a control diet (CD). Control mice (of the same strain as CPD mice) were housed in pairs and left unstressed in their home cage (UN). In all these mice, we evaluated body weight, different adipose depots, energy metabolism, caloric intake, and neuropeptide expression. RESULTS: CPD mice increased the intake of HFD and reduced body weight in the presence of enhanced lipid oxidation. Resting energy expenditure and interscapular brown adipose tissue (iBAT) were increased in CPD mice, whereas epididymal adipose tissue increased only in HFD-fed unstressed mice. Propiomelanocortin mRNA levels in hypothalamic arcuate nucleus increased only in HFD-fed unstressed mice. Oxytocin mRNA levels in the paraventricular nucleus and neuropeptide Y mRNA levels within the arcuate were increased only in CD-fed CPD mice. In the arcuate, CART was increased in HFD-fed UN mice and in CD-fed CPD mice, while HFD intake suppressed CART increase in defeated animals. In the basolateral amygdala, CART expression was increased only in CPD animals on HFD. CONCLUSIONS: CPD appears to uncouple the intake of HFD from energy homeostasis causing higher HFD intake, larger iBAT accumulation, increased energy expenditure and lipid oxidation, and lower body weight. Overall, the present study confirms the notion that the chronic activation of the stress response can be associated with metabolic disorders, altered energy homeostasis, and changes of orexigenic and anorexigenic signaling. These changes might be relevant to better understand the etiology of stress-induced obesity and eating disorders and might represent a valid therapeutic approach for the development of new therapies in this field.


Subject(s)
Diet, High-Fat , Energy Intake/physiology , Energy Metabolism/physiology , Animals , Body Weight , Italy , Leptin/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity
13.
Int J Mol Sci ; 19(7)2018 Jul 20.
Article in English | MEDLINE | ID: mdl-30036966

ABSTRACT

BACKGROUND: Organochlorine pesticides (OCPs) are widely distributed in the environment and their toxicity is mostly associated with the molecular mechanisms of endocrine disruption. Among OCPs, particular attention was focused on the effects of ß-hexaclorocyclohexane (ß-HCH), a widely common pollutant. A detailed epidemiological study carried out on exposed population in the "Valle del Sacco" found correlations between the incidence of a wide range of diseases and the occurrence of ß-HCH contamination. Taking into account the pleiotropic role of the protein signal transducer and activator of transcription 3 (STAT3), its function as a hub protein in cellular signaling pathways triggered by ß-HCH was investigated in different cell lines corresponding to tissues that are especially vulnerable to damage by environmental pollutants. MATERIALS AND METHODS: Human prostate cancer (LNCaP), human breast cancer (MCF-7 and MDA-MB 468), and human hepatoma (HepG2) cell lines were treated with 10 µM ß-HCH in the presence or absence of specific inhibitors for different receptors. All samples were subjected to analysis by immunoblotting and RT-qPCR. RESULTS AND CONCLUSIONS: The preliminary results allow us to hypothesize the involvement of STAT3, through both its canonical and non-canonical pathways, in response to ß-HCH. Moreover, we ascertained the role of STAT3 as a master regulator of energy metabolism via the altered expression and localization of HIF-1α and PKM2, respectively, resulting in a Warburg-like effect.


Subject(s)
Hexachlorocyclohexane/pharmacology , STAT3 Transcription Factor/metabolism , Energy Metabolism/drug effects , Female , Hep G2 Cells , Humans , MCF-7 Cells , Male , Signal Transduction/drug effects
14.
Biochim Biophys Acta ; 1830(3): 2671-82, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23671928

ABSTRACT

BACKGROUND: Green tea is a rich source of polyphenols, mainly catechins (flavanols), which significantly contribute to the beneficial health effects of green tea in the prevention and treatment of various diseases. In this study the effects of four green tea catechins on protein ERp57, also known as protein disulfide isomerase isoform A3 (PDIA3), have been investigated in an in vitro model. METHODS: The interaction of catechins with ERp57 was explored by fluorescence quenching and surface plasmon resonance techniques and their effect on ERp57 activities was investigated. RESULTS: A higher affinity was observed for galloylated cathechins, which bind close to the thioredoxin-like redox-sensitive active sites of the protein, with a preference for the oxidized form. The effects of these catechins on ERp57 properties were also investigated and a moderate inhibition of the reductase activity of ERp57 was observed as well as a strong inhibition of ERp57 DNA binding activity. CONCLUSIONS: Considering the high affinity of galloylated catechins for ERp57 and their capability to inhibit ERp57 binding to other macromolecular ligands, some effects of catechins interaction with this protein on eukaryotic cells may be expected. GENERAL SIGNIFICANCE: This study provides information to better understand the molecular mechanisms underlying the biological activities of catechins and to design new polyphenol-based ERp57-specific inhibitors.


Subject(s)
Catechin/analogs & derivatives , Catechin/chemistry , Protein Disulfide-Isomerases/chemistry , Catalytic Domain , DNA/chemistry , Enzyme Assays , Humans , Kinetics , Ligands , Models, Molecular , Oxidation-Reduction , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Spectrometry, Fluorescence , Structure-Activity Relationship , Surface Plasmon Resonance
15.
J Cell Biochem ; 114(11): 2461-70, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23696074

ABSTRACT

The disulfide isomerase ERp57 is a soluble protein mainly located in the endoplasmic reticulum, where it acts in the quality control of newly synthesized glycoproteins, in association with calreticulin and calnexin. It has been also detected in other cell compartments, such as the cytosol, the plasma membrane and the nucleus. In these locations it is implicated in various processes, participating in the rapid response to calcitriol, modulating the activity of STAT3 and being requested for the pre-apoptotic exposure of calreticulin on the plasma membrane. In the present work, the involvement of ERp57 in the activity of the EGF receptor was evaluated for the first time. EGFR is a tyrosine kinase receptor, which is able to activate numerous signaling cascades, leading to cell proliferation and inhibition of apoptosis. In the MDA-MB-468 breast adenocarcinoma cells, which overexpress EGFR, ERp57 expression has been knocked down by siRNA and the effects on EGFR have been studied. ERp57 silencing did not affect EGFR protein expression, cell membrane exposure or EGF binding, whereas the internalization and the phosphorylation of the receptor were impaired. The implication of ERp57 in the activity of EGFR, whose upregulation is known to be associated with tumors, could be relevant for cancer therapy.


Subject(s)
ErbB Receptors/metabolism , Protein Disulfide-Isomerases/metabolism , Cell Line, Tumor , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Membrane/physiology , ErbB Receptors/genetics , Fluorescent Antibody Technique , Humans , Protein Disulfide-Isomerases/genetics , RNA Interference , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology
16.
Exp Cell Res ; 318(19): 2460-9, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22814252

ABSTRACT

Dystroglycan (DG) is an extracellular receptor composed of two subunits, α-DG and ß-DG, connected through the α-DG C-terminal domain and the ß-DG N-terminal domain. We report an alanine scanning of all DG cysteine residues performed on DG-GFP constructs overexpressed in 293-Ebna cells, demonstrating that Cys-669 and Cys-713, both located within the ß-DG N-terminal domain, are key residues for the DG precursor cleavage and trafficking, but not for the interaction between the two DG subunits. In addition, we have used immunprecipitation and confocal microscopy showing that ERp57, a member of the disulfide isomerase family involved in glycoprotein folding, is associated and colocalizes immunohistochemically with ß-DG in the ER and at the plasma membrane of 293-Ebna cells. The ß-DG-ERp57 complex also included α-DG. DG mutants, unable to undergo the precursor cleavage, were still associated to ERp57. ß-DG and ERp57 were also co-immunoprecipitated in rat heart and kidney tissues. In vitro, a mutant ERp57, mimicking the reduced form of the wild-type protein, interacts directly with the recombinant N-terminal domain of both α-DG and ß-DG with apparent dissociation constant values in the micromolar range. ERp57 is likely to be involved in the DG processing/maturation pathway, but its association to the mature DG complex might also suggest some further functional role that needs to be investigated.


Subject(s)
Dystroglycans/metabolism , Protein Disulfide-Isomerases/metabolism , Animals , Cell Membrane/genetics , Cell Membrane/metabolism , Cells, Cultured , Dystroglycans/genetics , Glycosylation , HEK293 Cells , Heart/physiology , Humans , Kidney/metabolism , Kidney/physiology , Mutation , Protein Binding/genetics , Protein Binding/physiology , Protein Structure, Tertiary , Protein Subunits , Protein Transport/physiology , Rats
17.
Int J Mol Sci ; 14(6): 11259-76, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23759982

ABSTRACT

Extending our previous observations, we have shown on HaCat cells that melatonin, at ~10-9 M concentration, transiently raises not only the expression of the neuronal nitric oxide synthase (nNOS) mRNA, but also the nNOS protein synthesis and the nitric oxide oxidation products, nitrite and nitrate. Interestingly, from the cell bioenergetic point of view, the activated NO-related chemistry induces a mild decrease of the oxidative phosphorylation (OXPHOS) efficiency, paralleled by a depression of the mitochondrial membrane potential. The OXPHOS depression is apparently balanced by glycolysis. The mitochondrial effects described have been detected only at nanomolar concentration of melatonin and within a time window of a few hours' incubation; both findings compatible with the melatonin circadian cycle.


Subject(s)
Circadian Rhythm , Melatonin/metabolism , Mitochondria/metabolism , Nitric Oxide/metabolism , Adenosine Triphosphate/biosynthesis , Cell Line , Cell Respiration/drug effects , Circadian Rhythm/drug effects , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Lactates/metabolism , Melatonin/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Models, Biological , Nitric Oxide Synthase Type I/metabolism , Oxidative Phosphorylation/drug effects , Time Factors
18.
IUBMB Life ; 64(3): 251-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22271455

ABSTRACT

A novel role of melatonin was unveiled, using immortalized human keratinocyte cells (HaCaT) as a model system. Within a time window compatible with its circadian rhythm, melatonin at nanomolar concentration raised both the expression level of the neuronal nitric oxide synthase mRNA and the nitric oxide oxidation products, nitrite and nitrate. On the same time scale, a depression of the mitochondrial membrane potential was detected together with a decrease of the oxidative phosphorylation efficiency, compensated by glycolysis as testified by an increased production of lactate. The melatonin concentration, ∼ nmolar, inducing the bioenergetic effects and their time dependence, both suggest that the observed nitric oxide-induced mitochondrial changes might play a role in the metabolic pathways characterizing the circadian melatonin chemistry.


Subject(s)
Antioxidants/pharmacology , Energy Metabolism/drug effects , Keratinocytes/drug effects , Keratinocytes/enzymology , Melatonin/pharmacology , Nitric Oxide Synthase Type I/metabolism , Adenosine Triphosphate/metabolism , Blotting, Western , Cells, Cultured , Humans , Keratinocytes/cytology , Lactic Acid/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/genetics , Nitrites/metabolism , Oxidation-Reduction , Oxidative Phosphorylation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction
19.
PLoS Genet ; 5(10): e1000670, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19798443

ABSTRACT

Heterochromatin Protein 1 (HP1a) is a well-known conserved protein involved in heterochromatin formation and gene silencing in different species including humans. A general model has been proposed for heterochromatin formation and epigenetic gene silencing in different species that implies an essential role for HP1a. According to the model, histone methyltransferase enzymes (HMTases) methylate the histone H3 at lysine 9 (H3K9me), creating selective binding sites for itself and the chromodomain of HP1a. This complex is thought to form a higher order chromatin state that represses gene activity. It has also been found that HP1a plays a role in telomere capping. Surprisingly, recent studies have shown that HP1a is present at many euchromatic sites along polytene chromosomes of Drosophila melanogaster, including the developmental and heat-shock-induced puffs, and that this protein can be removed from these sites by in vivo RNase treatment, thus suggesting an association of HP1a with the transcripts of many active genes. To test this suggestion, we performed an extensive screening by RIP-chip assay (RNA-immunoprecipitation on microarrays), and we found that HP1a is associated with transcripts of more than one hundred euchromatic genes. An expression analysis in HP1a mutants shows that HP1a is required for positive regulation of these genes. Cytogenetic and molecular assays show that HP1a also interacts with the well known proteins DDP1, HRB87F, and PEP, which belong to different classes of heterogeneous nuclear ribonucleoproteins (hnRNPs) involved in RNA processing. Surprisingly, we found that all these hnRNP proteins also bind heterochromatin and are dominant suppressors of position effect variegation. Together, our data show novel and unexpected functions for HP1a and hnRNPs proteins. All these proteins are in fact involved both in RNA transcript processing and in heterochromatin formation. This suggests that, in general, similar epigenetic mechanisms have a significant role on both RNA and heterochromatin metabolisms.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Drosophila melanogaster/metabolism , Euchromatin/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , RNA/metabolism , Up-Regulation , Animals , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Drosophila melanogaster/chemistry , Drosophila melanogaster/genetics , Euchromatin/genetics , Gene Expression , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Protein Binding , RNA/chemistry , RNA/genetics , RNA Processing, Post-Transcriptional , RNA Stability
20.
Microsc Microanal ; 18(4): 829-39, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22697480

ABSTRACT

This study highlights the role of specific outer bacterial structures, such as the glycocalix, in calcium carbonate crystallization in vitro. We describe the formation of calcite crystals by extracellular polymeric materials, such as exopolysaccharides (EPS) and capsular polysaccharides (CPS) isolated from Bacillus firmus and Nocardia calcarea. Organic matrices were isolated from calcifying bacteria grown on synthetic medium--in the presence or absence of calcium ions--and their effect on calcite precipitation was assessed. Scanning electron microscopy observations and energy dispersive X-ray spectrometry analysis showed that CPS and EPS fractions were involved in calcium carbonate precipitation, not only serving as nucleation sites but also through a direct role in crystal formation. The utilization of different synthetic media, with and without addition of calcium ions, influenced the biofilm production and protein profile of extracellular polymeric materials. Proteins of CPS fractions with a molecular mass between 25 and 70 kDa were overexpressed when calcium ions were present in the medium. This higher level of protein synthesis could be related to the active process of bioprecipitation.


Subject(s)
Bacillus/chemistry , Bacillus/metabolism , Calcium Carbonate/chemistry , Nocardia/chemistry , Nocardia/metabolism , Polysaccharides, Bacterial/metabolism , Bacillus/ultrastructure , Bacterial Proteins/metabolism , Calcium/metabolism , Calcium Carbonate/metabolism , Crystallization , Microscopy, Electron, Scanning , Nocardia/ultrastructure , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/isolation & purification , Protein Biosynthesis , Spectrometry, X-Ray Emission
SELECTION OF CITATIONS
SEARCH DETAIL